Foot-and-mouth disease virus (FMDV)

口蹄疫病毒 ( FMDV )
  • 文章类型: Journal Article
    背景:口蹄疫(FMD)是一种影响偶蹄动物的破坏性疾病,这导致受影响国家和地区的重大经济损失。目前,有明显倾向于利用纳米粒子作为创新疫苗开发的强大平台。因此,这项研究开发了一种基于铁蛋白的纳米颗粒(FNP)疫苗,该疫苗在FNP表面上显示口蹄疫病毒(FMDV)VP1(aa140-158)的中和表位,并评估了这些FNP在小鼠和豚鼠模型中的免疫原性和保护功效,为开发潜在的FMD疫苗提供了策略。
    结果:本研究表达了重组蛋白Hpf,HPF-NE和HPF-T34E通过大肠杆菌表达系统。结果表明,重组蛋白Hpf,Hpf-NE和Hpf-T34E可以有效地组装成纳米颗粒。随后,我们评估了Hpf的免疫原性,小鼠Hpf-NE和Hpf-T34E蛋白,以及Hpf-T34E蛋白在豚鼠中的免疫原性和保护性。小鼠试验成果显示Hpf-T34E组的免疫效力年夜于Hpf-NE组。用Hpf-T34E免疫的豚鼠的结果表明,免疫效力与FMD灭活疫苗(IV)的免疫原性基本一致,并且可以在豚鼠中提供针对FMDV攻击的部分保护。
    结论:Hpf-T34E纳米颗粒作为抗口蹄疫亚单位疫苗候选物较好的选择,在FMDV感染的模型动物中提供有效的保护。基于FNP的疫苗表现出优异的安全性和免疫原性,因此代表了继续开发高效和安全的FMD疫苗的有希望的策略。
    BACKGROUND: Foot-and-mouth disease (FMD) is a devastating disease affecting cloven-hoofed animals, that leads to significant economic losses in affected countries and regions. Currently, there is an evident inclination towards the utilization of nanoparticles as powerful platforms for innovative vaccine development. Therefore, this study developed a ferritin-based nanoparticle (FNP) vaccine that displays a neutralizing epitope of foot-and-mouth disease virus (FMDV) VP1 (aa 140-158) on the surface of FNP, and evaluated the immunogenicity and protective efficacy of these FNPs in mouse and guinea pig models to provide a strategy for developing potential FMD vaccines.
    RESULTS: This study expressed the recombinant proteins Hpf, HPF-NE and HPF-T34E via an E. coli expression system. The results showed that the recombinant proteins Hpf, Hpf-NE and Hpf-T34E could be effectively assembled into nanoparticles. Subsequently, we evaluated the immunogenicity of the Hpf, Hpf-NE and Hpf-T34E proteins in mice, as well as the immunogenicity and protectiveness of the Hpf-T34E protein in guinea pigs. The results of the mouse experiment showed that the immune efficacy in the Hpf-T34E group was greater than the Hpf-NE group. The results from guinea pigs immunized with Hpf-T34E showed that the immune efficacy was largely consistent with the immunogenicity of the FMD inactivated vaccine (IV) and could confer partial protection against FMDV challenge in guinea pigs.
    CONCLUSIONS: The Hpf-T34E nanoparticles stand out as a superior choice for a subunit vaccine candidate against FMD, offering effective protection in FMDV-infected model animals. FNP-based vaccines exhibit excellent safety and immunogenicity, thus representing a promising strategy for the continued development of highly efficient and safe FMD vaccines.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    为了研制出安全有效的口蹄疫广谱疫苗,在这里,我们基于三种不同的FMDV拓扑类型开发了重组FMD多表位三价疫苗.通过在猪中的免疫效力评价疫苗的效力。结果表明,抗原不低于25μg的疫苗通过一级加强方案引发FMDV血清型O特异性抗体和中和抗体,并为猪提供免疫保护。更重要的是,该疫苗不仅引发了针对FMDV三种不同拓扑类型的相同水平的中和抗体,但也提供了完全保护猪免受三种相应的病毒攻击。在整个实验过程中,完全受保护的猪都无法产生抗3ABC抗体,这意味着疫苗可以提供无菌免疫。该疫苗可引发持久的高水平抗体,并在免疫后六个月内有效保护猪免受毒力攻击。因此,我们认为这种疫苗将来可能用于FMD的预防和控制。
    In order to develop a safe and effective broad-spectrum vaccine for foot-and-mouth disease (FMDV), here, we developed a recombinant FMD multiple-epitope trivalent vaccine based on three distinct topotypes of FMDV. Potency of the vaccine was evaluated by immune efficacy in pigs. The results showed that the vaccine with no less than 25 μg of antigen elicited FMDV serotype O specific antibodies and neutralization antibodies by primary-booster regime, and offered immune protection to pigs. More importantly, the vaccine elicited not only the same level of neutralization antibodies against the three distinct topotypes of FMDV, but also provided complete protection in pigs from the three corresponding virus challenge. None of the fully protected pigs were able to generate anti-3ABC antibodies throughout the experiment, which implied the vaccine can offer sterilizing immunity. The vaccine elicited lasting-long high-level antibodies and effectively protected pigs from virulent challenge within six months of immunization. Therefore, we consider that this vaccine may be used in the future for the prevention and control of FMD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    内部核糖体进入位点(IRES)元件构成顺式作用RNA调节序列,以不依赖帽的方式募集核糖体起始复合物,由各种RNA结合蛋白和IRES反式作用因子辅助。口蹄疫病毒(FMDV)含有功能性IRES元件,并利用该元件颠覆宿主翻译机制。我们的研究发现了一种新机制,其中RALY,属于RNA结合蛋白的异质核核糖核蛋白(hnRNP)家族的成员,通过其RNA识别基序残基与FMDVIRES的结构域3结合。这种相互作用通过抑制IRES驱动的翻译导致FMDV复制的下调。此外,我们的发现表明,RALY对FMDV复制的抑制作用不是归因于FMDVIRES介导的翻译起始复合物的组装,而是归因于与40S核糖体结合后80S核糖体复合物形成的障碍。相反,FMDV的3Cpro通过泛素-蛋白酶体途径抵消RALY介导的抑制。因此,这些结果表明,RALY,作为一种新型的关键IRES结合蛋白,通过阻断80S核糖体的形成来抑制FMDV的复制,提供对病毒如何招募和操纵宿主因素的更深入的了解。重要性由IRES元件驱动的FMDV基因组RNA的翻译是病毒感染的关键步骤。许多宿主蛋白被劫持以调节FMDVIRES依赖性翻译,但调控机制尚不清楚。这里,我们首次报道细胞RALY特异性地与FMDV的IRES相互作用,并通过阻断FMDVIRES上的80S核糖体组装来负调控病毒复制。相反,RALY介导的抑制通过泛素-蛋白酶体途径被病毒3C蛋白酶拮抗。这些结果将有助于进一步了解病毒感染过程中病毒与宿主的相互作用和翻译控制。
    The internal ribosome entry site (IRES) element constitutes a cis-acting RNA regulatory sequence that recruits the ribosomal initiation complex in a cap-independent manner, assisted by various RNA-binding proteins and IRES trans-acting factors. Foot-and-mouth disease virus (FMDV) contains a functional IRES element and takes advantage of this element to subvert host translation machinery. Our study identified a novel mechanism wherein RALY, a member of the heterogeneous nuclear ribonucleoproteins (hnRNP) family belonging to RNA-binding proteins, binds to the domain 3 of FMDV IRES via its RNA recognition motif residue. This interaction results in the downregulation of FMDV replication by inhibiting IRES-driven translation. Furthermore, our findings reveal that the inhibitory effect exerted by RALY on FMDV replication is not attributed to the FMDV IRES-mediated assembly of translation initiation complexes but rather to the impediment of 80S ribosome complex formation after binding with 40S ribosomes. Conversely, 3Cpro of FMDV counteracts RALY-mediated inhibition by the ubiquitin-proteasome pathway. Therefore, these results indicate that RALY, as a novel critical IRES-binding protein, inhibits FMDV replication by blocking the formation of 80S ribosome, providing a deeper understanding of how viruses recruit and manipulate host factors.
    OBJECTIVE: The translation of FMDV genomic RNA driven by IRES element is a crucial step for virus infections. Many host proteins are hijacked to regulate FMDV IRES-dependent translation, but the regulatory mechanism remains unknown. Here, we report for the first time that cellular RALY specifically interacts with the IRES of FMDV and negatively regulates viral replication by blocking 80S ribosome assembly on FMDV IRES. Conversely, RALY-mediated inhibition is antagonized by the viral 3C protease by the ubiquitin-proteasome pathway. These results would facilitate further understanding of virus-host interactions and translational control during viral infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    口蹄疫(FMD)是一种引起重大经济损失的急性人畜共患病。迫切需要能够刺激有效保护性免疫应答的疫苗。在这项研究中,使用强大的插件和展示SpyTag/SpyCatcher系统,将血清型A和OFMD病毒(FMDV)的大肠杆菌衍生的重组VP1与热稳定的支架lumazine合酶(LS)或耐热拟杆菌封装蛋白(QtEnc)缀合,以产生多聚纳米疫苗。这些纳米疫苗在接种的小鼠中诱导高度有效的抗体应答。第一次免疫后第14天,抗体滴度比单体抗原高约100倍。两种疫苗均诱导高和长期IgG抗体产生。此外,QtEnc-VP1纳米疫苗诱导的抗体滴度高于LS-VP1纳米疫苗。纳米疫苗还诱导Th1偏向的免疫应答和更高水平的中和抗体。这些数据表明,通过将VP1与热稳定的支架缀合而产生的FMDV纳米疫苗是低资源地区FMDV控制的高度免疫原性和理想候选物。
    Foot-and-mouth disease (FMD) is an acute zoonosis causes significant economic losses. Vaccines able to stimulate efficient protective immune responses are urgently needed. In this study, Escherichia coli-derived recombinant VP1 of serotype A and O FMD virus (FMDV) was conjugated to thermostable scaffold lumazine synthase (LS) or Quasibacillus thermotolerans encapsulin (QtEnc) using a robust plug-and-display SpyTag/SpyCatcher system to generate multimeric nanovaccines. These nanovaccines induced highly potent antibody responses in vaccinated mice. On day 14 after the first immunisation, antibody titres were approximately 100 times higher than those of monomer antigens. Both vaccines induced high and long-term IgG antibody production. Moreover, the QtEnc-VP1 nanovaccine induced higher antibody titres than the LS-VP1 nanovaccine. The nanovaccines also induced Th1-biased immune responses and higher levels of neutralising antibodies. These data indicated that FMDV nanovaccines generated by conjugating VP1 with a thermostable scaffold are highly immunogenic and ideal candidates for FMDV control in low-resource areas.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: English Abstract
    瞬时表达是在哺乳动物细胞中表达口蹄疫病毒(FMDV)衣壳蛋白的主要方法。为了实现FMDV衣壳蛋白的稳定表达和病毒样颗粒(VLPs)在细胞中的高效组装,构建piggyBac(PB)转座子组成型表达质粒和PB转座子-四环素(Tet)诱导型表达载体。通过荧光蛋白测试质粒的功能。通过添加抗生素,组成型细胞池(C-WT,C-L127P)表达P12A3C(WT/L127P)基因和可诱导细胞池(I-WT,产生了表达P12A3C(WT/L127P)的I-L127P)基因。绿色荧光蛋白的基因,3C蛋白酶和反向四环素反式激活剂(rtTA)整合到染色体上,荧光观察和PCR检测证实了这一点。细胞池I-L127P具有更强的衣壳蛋白和VLP生产能力,通过蛋白质印迹和酶联免疫吸附测定(ELISA)证实,分别。总之,首次报道了诱导FMDV衣壳蛋白的染色体表达,这可能有助于哺乳动物生产FMDVVLP疫苗的技术过程以及其他蛋白质的哺乳动物诱导型表达系统的构建。
    Transient expression is the major method to express foot-and-mouth disease virus (FMDV) capsid proteins in mammalian cells. To achieve stable expression of FMDV capsid proteins and efficient assembly of virus like particles (VLPs) in cells, the plasmids of piggyBac (PB) transposon-constitutive expression and PB transposon-tetracycline (Tet) inducible expression vectors were constructed. The function of the plasmids was tested by fluorescent proteins. By adding antibiotics, the constitutive cell pools (C-WT, C-L127P) expressing P12A3C (WT/L127P) genes and the inducible cell pools (I-WT, I-L127P) expressing P12A3C (WT/L127P) genes were generated. The genes of green fluorescent protein, 3C protease and reverse tetracycline transactivator (rtTA) were integrated into chromosome, which was confirmed by fluorescence observation and PCR testing. The cell pool I-L127P has a stronger production capacity of capsid proteins and VLPs, which was confirmed by Western blotting and enzyme linked immunosorbent assay (ELISA), respectively. In conclusion, inducing the chromosomal expression of FMDV capsid proteins was firstly reported, which may facilitate the technical process of mammalian production of FMDV VLPs vaccine and the construction of mammalian inducible expression systems for other proteins.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    口蹄疫病毒(FMDV)已经开发了各种策略来拮抗宿主的先天免疫。FMDVLpro和3Cpro通过不同的机制干扰I型IFN。FMDV的结构蛋白VP3降解Janus激酶1以抑制IFN-γ信号转导。FMDV的非结构蛋白是否参与抑制II型IFN信号通路尚不清楚。在这项研究中,研究表明,感染建立后,FMDV复制对IFN-γ治疗具有抗性,并且FMDV抑制II型IFN诱导的IFN-γ刺激基因(ISG)的表达。我们还首次显示FMDV非结构蛋白3C通过阻断STAT1核易位来拮抗IFN-γ刺激的JAK-STAT信号通路。3Cpro表达显着降低了ISGs转录水平和回文γ激活序列(GAS)启动子活性,在不影响蛋白质水平的情况下,酪氨酸磷酸化,和STAT1的同源二聚化。最后,我们提供的证据表明,3C蛋白酶活性在降解KPNA1中起着至关重要的作用,因此抑制了ISGsmRNA和GAS启动子的活性。我们的结果揭示了FMDV非结构蛋白拮抗宿主II型IFN信号的新机制。
    Foot-and-mouth disease virus (FMDV) has developed various strategies to antagonize the host innate immunity. FMDV Lpro and 3Cpro interfere with type I IFNs through different mechanisms. The structural protein VP3 of FMDV degrades Janus kinase 1 to suppress IFN-γ signaling transduction. Whether non-structural proteins of FMDV are involved in restraining type II IFN signaling pathways is unknown. In this study, it was shown that FMDV replication was resistant to IFN-γ treatment after the infection was established and FMDV inhibited type II IFN induced expression of IFN-γ-stimulated genes (ISGs). We also showed for the first time that FMDV non-structural protein 3C antagonized IFN-γ-stimulated JAK-STAT signaling pathway by blocking STAT1 nuclear translocation. 3Cpro expression significantly reduced the ISGs transcript levels and palindromic gamma-activated sequences (GAS) promoter activity, without affecting the protein level, tyrosine phosphorylation, and homodimerization of STAT1. Finally, we provided evidence that 3C protease activity played an essential role in degrading KPNA1 and thus inhibited ISGs mRNA and GAS promoter activities. Our results reveal a novel mechanism by which an FMDV non-structural protein antagonizes host type II IFN signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    用完整(146S)口蹄疫病毒(FMDV)颗粒接种疫苗用于控制口蹄疫。然而,146S颗粒容易解离成稳定的五聚体12S颗粒,其免疫原性较低。我们先前分离了几种特异性结合146S或12S颗粒的单结构域抗体片段(VHH)。这些颗粒特异性VHH是疫苗质量控制的极好工具。在这项研究中,我们通过竞争ELISA绘制了这些VHH识别的抗原位点,病毒中和,和表位的胰蛋白酶敏感性。我们包括两种先前描述的12S特异性(mAb13A6)或146S特异性(mAb9)的单克隆抗体(mAb)。虽然两者都是12S特定的,发现VHHM3F和mAb13A6结合独立的抗原位点。M3F识别非中和和胰蛋白酶不敏感位点,而mAb13A6识别胰蛋白酶敏感的VP2N末端。Asia1146S特异性位点是胰蛋白酶敏感的,中和也被VHHM8F识别,这表明它涉及VP1GH循环。A型146S特异性VHH识别两个独立的抗原位点,它们也中和但胰蛋白酶不敏感。主要位点通过与FMDV复合的两个广泛的菌株反应性146S特异性VHHs的交联质谱(XL-MS)进一步作图。表位位于二十面体病毒3D结构的2倍和3倍对称轴附近,主要在VP2和VP3上,与较早鉴定的mAb9位点重叠。由于表位位于单个12S五聚体上,146S特异性不能通过由于12S五聚体解离而分裂的表位来解释。在较早的研究中,与O型FMDV复合的146S特异性VHHM170的低温EM结构得到了解决。据报道146S特异性是由12S和146S颗粒中该表位的构象改变引起的。这种机制可能也解释了XL-MS定位的两种A型VHH的146S特异性结合,因为它们的表位与M170识别的表位重叠。令人惊讶的是,146S四级结构内部的残基也与VHH交联。这可能反映了溶液中的颗粒灵活性。病毒-抗体相互作用的分子研究有助于进一步优化疫苗并改善其质量控制。
    Vaccination with intact (146S) foot-and-mouth disease virus (FMDV) particles is used to control FMD. However, 146S particles easily dissociate into stable pentameric 12S particles which are less immunogenic. We earlier isolated several single-domain antibody fragments (VHHs) that specifically bind either 146S or 12S particles. These particle-specific VHHs are excellent tools for vaccine quality control. In this study we mapped the antigenic sites recognized by these VHHs by competition ELISAs, virus neutralization, and trypsin sensitivity of epitopes. We included two previously described monoclonal antibodies (mAbs) that are either 12S specific (mAb 13A6) or 146S specific (mAb 9). Although both are 12S specific, the VHH M3F and mAb 13A6 were found to bind independent antigenic sites. M3F recognized a non-neutralizing and trypsin insensitive site whereas mAb 13A6 recognized the trypsin sensitive VP2 N-terminus. The Asia1 146S-specific site was trypsin sensitive, neutralizing and also recognized by the VHH M8F, suggesting it involves the VP1 GH-loop. The type A 146S-specific VHHs recognized two independent antigenic sites that are both also neutralizing but trypsin insensitive. The major site was further mapped by cross-linking mass spectrometry (XL-MS) of two broadly strain reactive 146S-specific VHHs complexed to FMDV. The epitopes were located close to the 2-fold and 3-fold symmetry axes of the icosahedral virus 3D structure, mainly on VP2 and VP3, overlapping the earlier identified mAb 9 site. Since the epitopes were located on a single 12S pentamer, the 146S specificity cannot be explained by the epitope being split due to 12S pentamer dissociation. In an earlier study the cryo-EM structure of the 146S-specific VHH M170 complexed to type O FMDV was resolved. The 146S specificity was reported to be caused by an altered conformation of this epitope in 12S and 146S particles. This mechanism probably also explains the 146S-specific binding by the two type A VHHs mapped by XL-MS since their epitopes overlapped with the epitope recognized by M170. Surprisingly, residues internal in the 146S quaternary structure were also cross-linked to VHH. This probably reflects particle flexibility in solution. Molecular studies of virus-antibody interactions help to further optimize vaccines and improve their quality control.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    蛋白质SUMO化代表重要的细胞过程,其调节许多宿主蛋白质以及许多侵袭性病毒蛋白质的活性。口蹄疫病毒(FMDV)是发现的第一种动物病毒。然而,SUMO化是否在FMDV感染过程中发生以及在FMDV发病机制中的作用尚未研究。在本研究中,我们证明了SUMO化通过小干扰RNA(siRNA)转染抑制FMDV的复制与SUMO化的药物抑制,这进一步证实了在3C蛋白酶突变的SUMO化缺陷型FMDV的病毒复制增加,亚代谢的目标。此外,我们提供了四个赖氨酸残基的证据,Lys-51,-54,-110和-159一起工作,将SUMO化赋予FMDV3C蛋白酶,这可能使FMDV3C的SUMO化更稳定,并提高宿主抑制FMDV复制的机会。这是首次报道可以通过SUMO化选择性修饰四个赖氨酸残基。最后,我们表明SUMO化减弱了切割能力,干扰素信号通路的抑制作用,和FMDV3C的蛋白质稳定性,这似乎与FMDV复制的减少有关。一起来看,我们的实验结果描述了一种新的细胞调节事件,该事件通过3C蛋白酶的SUMO化显著限制了FMDV的复制。重要性口蹄疫是偶蹄动物的高度传染性和经济上重要的疾病。SUMOylation,一种小的泛素样蛋白与多种底物蛋白的共价连接,已成为重要的翻译后修饰,在多种生物过程中起着多种作用。在这项研究中,发现FMDV3C的四个赖氨酸残基被SUMO化选择性修饰。此外,我们证明了SUMO化通过多种机制减弱FMDV3C的功能,包括卵裂能力,干扰素信号通路的抑制作用,和蛋白质的稳定性,which,反过来,导致FMDV复制减少。我们的发现表明,FMDV3C的SUMO化可以作为抵抗FMDV复制的宿主细胞防御。对驱动这一过程的细胞和分子机制的进一步理解应该提供新的见解,以设计一种有效的策略来控制FMDV在动物中的传播。
    Protein SUMOylation represents an important cellular process that regulates the activities of numerous host proteins as well as of many invasive viral proteins. Foot-and-mouth disease virus (FMDV) is the first animal virus discovered. However, whether SUMOylation takes place during FMDV infection and what role it plays in FMDV pathogenesis have not been investigated. In the present study, we demonstrated that SUMOylation suppressed FMDV replication by small interfering RNA (siRNA) transfection coupled with pharmaceutical inhibition of SUMOylation, which was further confirmed by increased virus replication for SUMOylation-deficient FMDV with mutations in 3C protease, a target of SUMOylation. Moreover, we provided evidence that four lysine residues, Lys-51, -54, -110, and -159, worked together to confer the SUMOylation to the FMDV 3C protease, which may make SUMOylation of FMDV 3C more stable and improve the host\'s chance of suppressing the replication of FMDV. This is the first report that four lysine residues can be alternatively modified by SUMOylation. Finally, we showed that SUMOylation attenuated the cleavage ability, the inhibitory effect of the interferon signaling pathway, and the protein stability of FMDV 3C, which appeared to correlate with a decrease in FMDV replication. Taken together, the results of our experiments describe a novel cellular regulatory event that significantly restricts FMDV replication through the SUMOylation of 3C protease. IMPORTANCE FMD is a highly contagious and economically important disease in cloven-hoofed animals. SUMOylation, the covalent linkage of a small ubiquitin-like protein to a variety of substrate proteins, has emerged as an important posttranslational modification that plays multiple roles in diverse biological processes. In this study, four lysine residues of FMDV 3C were found to be alternatively modified by SUMOylation. In addition, we demonstrated that SUMOylation attenuated FMDV 3C function through multiple mechanisms, including cleavage ability, the inhibitory effect of the interferon signaling pathway, and protein stability, which, in turn, resulted in a decrease of FMDV replication. Our findings indicate that SUMOylation of FMDV 3C serves as a host cell defense against FMDV replication. Further understanding of the cellular and molecular mechanisms driving this process should offer novel insights to design an effective strategy to control the dissemination of FMDV in animals.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Receptors interaction protein 2 (RIP2) is a specific adaptor molecule in the downstream of NOD2. The role of RIP2 during foot-and-mouth disease virus (FMDV) infection remains unknown. Here, our results showed that RIP2 inhibited FMDV replication and played an important role in the activation of IFN-β and NF-ĸB signal pathways during FMDV infection. FMDV infection triggered RIP2 transcription, while it reduced the expression of RIP2 protein. Detailed analysis showed that FMDV 2B, 2C, 3Cpro, and Lpro proteins were responsible for inducing the reduction of RIP2 protein. 3Cpro and Lpro are viral proteinases that can induce the cleavage or reduction of many host proteins and block host protein synthesis. The carboxyl terminal 105-114 and 135-144 regions of 2B were essential for reduction of RIP2. Our results also showed that the N terminal 1-61 region of 2C were essential for the reduction of RIP2. The 2C-induced reduction of RIP2 was dependent on inducing the reduction of poly(A)-binding protein 1 (PABPC1). The interaction between RIP2 and 2C was observed in the context of viral infection, and the residues 1-61 were required for the interaction. These data clarify novel mechanisms of reduction of RIP2 mediated by FMDV.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    The non-structural protein 3A of foot-and-mouth disease virus (FMDV) plays an important role in viral replication, virulence and determination of host range. Previously we identified genomic changes in gene encoding 3A protein between the attenuated ZBatt strain and its parental virulent strain during the attenuation process. However, the effects of changes in 3A protein on viral replication and infection of the rabbit-attenuated ZBatt virus during the attenuation process are poorly understood. In this study, a chimeric virus, rZBatt-3A, was constructed by introducing the 3A gene of virulent ZB virus into its attenuated vaccine ZB strain. Subsequently, the biological characteristics between rZBatt-3A and its parental virus (rZBatt) were compared. The relative expression level of four host cell proteins that interact with FMDV 3A were also analyzed. The results showed that the chimeric virus rZBatt-3A exhibited significantly different growth properties and plaque phenotypes from rZBatt in primary fetal bovine kidney (BK) cells. Cytopathic effect (CPE) of the rZBatt-3A was observed in BK cells with smaller plaque size, but CPE from the rZBatt could not be observed. The viral RNA replication was higher in rZBatt-3A-infected BK cells than in rZBatt-infected cells at 24 hpi (P < .05). In addition, the relative mRNA expression level of Ubiquilin 1 (UBQLN1) was significantly increased in rZBatt-3A-infected BK cells than in rZBatt-infected cells (P < .01) suggesting that UBQLN1 may be associated with 3A protein changes. Thus, the substitution of 3A protein altered the replication efficiency of attenuated ZB virus in bovine cells. Our data suggested that changes in 3A protein might be associated with the attenuation of ZB virus, which shed more lights in molecular mechanisms about attenuation of FMDV.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号